<<

0031-3998/06/5904-0094R PEDIATRIC RESEARCH Vol. 59, No. 4, Pt 2, 2006 Copyright © 2006 International Pediatric Research Foundation, Inc. Printed in U.S.A.

Adult -Derived Stem Cells for the : Implications for Pediatric Lung Diseases

TIMOTHY VAN HAAFTEN, AND BERNARD THE´ BAUD Department of Pediatrics, Division of Neonatology, Vascular Biology Research Group, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada

ABSTRACT: Bronchopulmonary dysplasia (BPD) and cystic fibro- the “stemness” of the stem cells used are all contentious sis (CF) are two common serious chronic respiratory disorders issues. Furthermore, stem research has been markedly without specific treatments affecting children. BPD is characterized slower in lung diseases due to the complexity of the lung. The by an arrest in alveolar growth in premature infants requiring respi- lung possesses numerous anatomical areas (upper airways, ratory support. CF is the most common fatal inherited genetic bronchioles, alveoli and their underlying capillaries) each of disorder characterized by abnormally thick mucus secretions, recur- which contains a unique cellular population with drastically rent infection and ultimately lung destruction. One commonality between these two diseases is the promise of utilizing stem cells different functions. therapeutically. Indeed, the use of exogenous cells to supplement the There are two common, serious pediatric lung diseases natural repair mechanisms or the possibility of genetic manipulation without current treatments that stem cells hold potential prom- before administration are appealing therapeutic options for ise for: bronchopulmonary dysplasia (BPD) and cystic fibrosis these diseases. Increasing attention has been focused on the use of (CF). BPD is the chronic lung disease of prematurity that adult bone marrow-derived stem cells (BMSC) to regenerate dam- follows ventilator support. BPD is characterized by an arrest aged organs such as the heart, the brain, and the liver. However, due in alveolar and vascular growth (1,2). The long-term conse- to the lung’s complexity as well as the low rate of cellular turnover quences of this halted lung growth are still unknown. There- within the lung, progress has been slower in this area compared with fore, the potential of a to restore or protect the the skin or liver. Initial work suggests that BMSC can engraft and alveolar epithelium is very attractive as this may reduce the differentiate into a variety of lung cells, but these findings have been morbidity and mortality associated with BPD. CF results from challenged recently. This article critically reviews the current ad- vances on the therapeutic use of stem cells for lung regeneration. mutations in the cystic fibrosis transmembrane conductance (Pediatr Res 59: 94R–99R, 2006) regulator (CFTR) gene and affects 1 in 3200 live births (3). There are five classes of gene defects in CF: 1) defective protein production; 2) aberrant intracellular protein trafficking; he current promise that stem cells hold for regen- 3) defective regulation of chloride transport; 4) reduced chlo- Teration is immense. The belief that once irreversibly ride transport function; 5) reduced protein expression. The damaged tissue can be restored to a normal functional capacity most common cause of CF occurs within class 2 (4). The is providing great optimism for novel treatments in many aberrant intracellular protein trafficking is caused by the de- diseases. However, this research is not without its controver- letion of an amino acid, commonly referred to as ⌬F508 (3,4). sies. There are ethical and moral issues surrounding the use of Pulmonary complications are typically the most serious and stem cells derived from . Recent research is starting to life-threatening manifestations of CF. Major advances in the unravel the potential for adult derived stem cells that was multidisciplinary management of CF have contributed to pro- initially described more than 20 y ago. While the initial results long the life span of affected patients to the mid 30s, but from numerous disease models outside the lung show great quality of life and other CF-related complications justify the potential for a cell replacement therapy, there is heated debate search for a cure. Stem cells have the potential to act as a over numerous concerns within the stem cell field. The mech- vehicle for corrective gene therapy, thereby restoring normal anism of action, the plasticity, the source, the phenotype, and function. Ultimately, lung stem cell therapy may also be

Received November 16, 2005; accepted December 14, 2005. Abbreviations: AEC, alveolar epithelial cell; BMSC, bone marrow-derived Correspondence: Bernard The´baud, M.D., Ph.D., Department of Pediatrics, Division stem cell; BPD, bronchopulmonary dysplasia; CF, cystic fibrosis; CFTR, of Neonatology, Vascular Biology Group, HMRC 407, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada, Phone: (780)-492-9479, Fax: (780)-492-3603: cystic fibrosis transmembrane conductance regulator; eGFP, enhanced green [email protected] fluorescent protein; ES cell, ; GFP, green fluorescent Dr. The´baud is supported by the Canada Foundation for Innovation, the Alberta Heart protein; HSC, ; MSC, ; SP and Stroke Foundation (H&SF), the Alberta Hertiage Foundation for Medical Research cell, sidepopulation cell; SP-C, surfactant protein C (AHFMR), the Canadian Institutes for Health Research (CIHR) and the Stollery Chil- dren’s Hospital Foundation. Tim van Haaften is supported by the CIHR-funded Torch (Tomorrow’s Research Cardiovascular Health Professionals) and MFN (Maternal-Fetal-Newborn Health) pro- grams. DOI: 10.1203/01.pdr.0000203550.50258.5a

94R STEM CELLS FOR LUNG DISEASES 95R applicable to other devastating lung disorders such as pulmo- heart, liver, brain, skeletal muscle, and vascular endothelium nary hypoplasia. (15). A true potential for adult derived stem cells lies with the This review will critically analyze the controversial litera- multipotent MSCs and these cells have been extensively used ture investigating the use of exogenously administered stem in lung stem cell research. This potential was first proposed in cells for lung diseases. the 1970s by Friedenstein et al. (16). This initial report showed that plastic adherent cells, previously believed to be DEFINITION OF STEM CELLS committed to a specific tissue type (i.e. bone marrow), had the ability to form cells within the osteogenic, chondrogenic, and Stem cells may be artificially classified into three catego- adipogenic lineages (16). There have been numerous reports ries: embryonic stem (ES) cells, adult bone marrow derived characterizing the phenotype of these MSCs in both humans stem cells (BMSC), and tissue progenitor cells (Table 1). ES (17,18) and mice (16,19). However, it was not until over 20 y cells are pluripotent cells derived from the inner cell mass of after Friedenstein’s first report that the true potential of these blastocyst-stage embryos (5). They are able to form tissue cells were made apparent. In 2001, Krause et al. reported that within each of the germ layers (the , , and a single bone-marrow derived stem cell had the ability to ectoderm) and have thus enormous therapeutic potential to engraft into numerous organs as well as having a “tremendous regenerate any damaged tissue; but ES cells are also mired in differentiative capacity” by adopting the phenotype of epithe- great controversy due to the present need of destroying an lial cells within the liver, lung, GI tract, and skin (20). to harvest pluripotent cells (6). Conversely, progeni- This initial report from Krause has led to extensive study of tor cells are thought to reside within a tissue and are stimu- these cells in the heart (21–24), liver (25–27), brain (28), lated for repair after injury. It is these tissue progenitor cells kidney (29), and endothelium (30). And while these initial that give the liver (the hepatic oval cell) its robust regenerative results show great promise in providing new therapies for ability (7,8). In the lung, type II alveolar epithelial cells (AEC) numerous diseases, these studies have also generated great are the putative distal progenitor cells responsible for repair controversy and highlight the discrepancies that exist within after injury (9,10). Side population (SP) cells, identified by adult stem cell research. These controversies include the their ability to efflux Hoechst dye, and tracheal epithelial basal source and phenotype of the stem cells used, the experimental cells, are putative progenitor cells in the upper airways (11). model of injury, the mechanism of effect, and the methods While the pluripotency of ES and further characterization of used to determine efficacy. While stem cell research in the resident lung stem cells (12,13) hold promise for novel ther- lung has been less prolific than in cardio-vascular or neuro- apeutic approaches for lung diseases, this review will focus on logic diseases, the same controversies apply and will be the adult bone-marrow derived stem cell and their exogenous discussed below. administration in various models of lung diseases. The bone marrow contains several population of primitive cells: 1) Hematopoeitic stem cells (HSCs); 2) mesenchymal STEM CELL THERAPIES TO PREVENT stem cells (MSCs); 3) endothelial stem/progenitor cells LUNG INJURY (EPC); 4) SP cells; and 5) multipotent adult progenitor cells The 2001 report from Krause et al. opened the door to the (MAPC). Among these, the HSCs are the best characterized of potential that there may be an ethically appealing option in the adult stem cells (14). HSCs are multipotent and have the using stem cells for tissue replacement. Based on their previ- ability to maintain or restore the mature circulating blood cells ous work showing the potential of CD34ϩ linϪ for regener- (erythrocytes and leukocytes). Recent evidence suggest that ating (27), Krause et al. extended their observa- bone-marrow derived cells have the ability to cross lineage tion to the lung. Myelo-ablation was induced in female mice barriers and generate differentiated tissue beyond their own by total body irradiation of 1050–1100 cGy (an otherwise tissue boundaries to form functional components of other lethal dose if the bone marrow is not reconstituted; in most tissues, expressing tissue-specific proteins in organs such as cases of irradiation the are also injured). CD34ϩ linϪ cells from male donor mice were administered intravenous Table 1. Terminology (IV) (20). Fluorescent in situ hybridization (FISH) to track the Y-chromosome showed 20% of AEC being derived from a Term Definition Example donor animal. Donor derived BMSC persisted up to 11 mo Totipotent Able to form any , Fertilized oocyte, post-transplant (20). includes the embryo and the trophoblast of the Consistent with this first observation, Kotton et al. reported 6 that plastic adherent MSCs (1–2 ϫ 10 injected IV) from Pluripotent Able to form cells within Embryonic stem cells transgenic mice overexpressing lacZ, allowing tracking of the any of the germ layers: cells via X-gal staining, were able to engraft and form type I the endoderm, ectoderm, AEC in experimental lung fibrosis induced by intratracheal and mesoderm Multipotent Able to differentiate into Mesenchymal stem cells, (IT) bleomycin (31). Bleomycin is one of the most extensively a select range of cell hematopoietic stem cells, studied and reproducible experimental models for lung fibro- types, heavily influenced certain tissue-specific stem sis. When bleomycin is given into the airway, it produces lung by the microenvironment cells, side-population epithelial injury, followed by an inflammatory response over cells several days, followed by lung fibrosis that eventually resolves 96R VAN HAAFTEN AND THE´ BAUD (32). Engrafted cells had morphologic features of type I AEC from the bleomycin-induced inflammation, as assessed by (i.e. flattened with ovoid nuclei bulging into alveolar lumen collagen deposition, and matrix metalloproteinase (MMP) and adjacent to type II AEC) and expressed type I AEC activation (40) as compared with animals that received MSCs specific markers such as Lycopersicon esculentum lectin and one week following bleomycin, indicating the importance of T1␣ (31). The proportion of donor derived MSCs contributing timing of stem cell therapy, as observed in myocardial infarct to the type I AEC population was not assessed. Interestingly (41). The low numbers of donor-derived cells engrafting the however, all bleomycin injured-mice (4/4) showed engraft- lung did not appear sufficient to account for the therapeutic ment when compared with PBS injected controls (2/9), sug- response, suggesting that donor stem cells may have other gesting that prior injury amplifies stem cell engraftment, as local effects. has been observed in other tissues (7). Furthermore, there Rojas et al. used a model in which before bleomycin were no donor-derived type II AECs, even in lungs harvested injury, the animals also underwent myelosuppression via a 1 and 2.5 d postinjection. In vitro, 10% of plastic adherent single administration of busulfan (42). Myelosuppression in- donor bone marrow cells cultured for one week expressed T1␣ creased the susceptibility to bleomycin injury, suggesting that and aquaporin 5, used as markers for type I AEC, (but are not an intact bone marrow serves to limit the extent of lung injury. solely confined to these cells) (33–35), but not surfactant After this suppression of the bone marrow, plastic adherent protein C (SP-C), a type II AEC specific marker (36) suggest- and immuno-depleted MSCs were given IV MSCs adminis- ing that cultured bone marrow cells can serve as type I AEC tration improved survival when compared with untreated an- precursors (31). imals. Furthermore, within the MSC administered group, These two reports form the prototypes for most of the there were a “substantial number” of donor cells within the experiments on BMSC in the lung that have followed. In lungs 2 wk following bleomycin insult with characteristics 2002, Theise et al. reported that donor-markers for either of AEC I and II, fibroblasts, and endothelial cells. In the whole bone marrow or CD34ϩ linϪ cells were detected in bleomycinϩbusulfan group, 29% of cells were derived from 14% of type II AEC after 1200 cGy total body irradiation (37). donor MSCs. Interestingly, circulating levels of granulocyte- Engraftment was still present 6 months later (for total bone macrophage colony-stimulating factor (GM-CSF) and granu- marrow injection) and 8 mo later for CD34ϩ linϪ cells, locyte colony stimulating factor (G-CSF) (factors known to detectable by FISH. In another study, administration of stimulate stem cell mobilization from the bone marrow), were MAPC to nonobese diabetic/severe combined immunodefi- higher in bleomycin-injured animals receiving MSC. This ciency (NOD/SCID) mice following mild irradiation of 250 suggests that, besides lung engraftment, humoral factors may cGy accounted for 4% of the alveolar epithelium assessed by also contribute to stem cell-induced tissue protection. This is ␤-galactosidase immunofluorescence (38). In another elegant consistent with the greater protection conferred by stem cell study, Abe et al. generated parabiotic mice by surgically transplant in bone marrow sufficient (no busulfan) animals. joining green fluorescent protein (GFP) transgenic mice and Preliminary data in oxygen-induced experimental BPD in wild-type littermates (39). These mice develop a common newborn rats also suggest a protective role of MSC. Rats circulation (approximately 50% green cells in blood) by 2 wk exposed to 95% oxygen from birth to 14 d postnatal during the after surgery. The wild-type mouse was either uninjured or alveolar period of lung development have an irreversible irradiated or received IT elastase (a well accepted emphysema arrest in lung development (43). In this preliminary study, model) or the combination of radiation with IT elastase injec- MSC were given IT, a situation that is clinically relevant since tion. Radiation or the combination of radiation with elastase premature infants at risk are routinely given surfactant via this significantly increased the proportion of bright green cells in route and which has been used successfully with other exper- the lungs of wild-type mice. These cells resembled morpho- imental therapies (44). CFSE (a green fluorescent marker)- logically, interstitial monocytes/macrophages, subepithelial fi- labeled MSC given at 4 d postnatal in hyperoxic-exposed broblast-like interstitial cells, and type I AEC. Approximately animals prevented stunted alveolar growth, engrafted and 5 to 20% of lung fibroblasts primary cultured from injured expressed the type II AEC specific marker SP-C (unpublished wild-type mice expressed GFP. data). Ortiz et al. used plastic adherent and purified, magnetic These studies demonstrate the potential of BMSC to home bead immunodepleted MSCs from male bleomycin-resistant to the injured lung, engraft, and adopt the phenotype of one or BALB/c mice expressing CD34, CD45, and CD11b in bleo- many of the lung cells. These reports also illustrate the mycin (4 U/kg)-induced pulmonary fibrosis in female bleo- numerous methods that exist in culturing and selecting BMSC mycin-sensitive C57BL/6 recipients (40). The purified MSCs (Table 2). The differences in experimental design and the (5 ϫ 105) were injected into the jugular vein. Lung engraft- numerous techniques to determine treatment efficacy (engraft- ment quantified by real-time PCR showed that male DNA ment, differentiation, improvement in lung histology) further accounted for 2.21 ϫ 10Ϫ5% of the total lung DNA in render the interpretation of these results difficult. control-treated mice but prior injury with bleomycin increased Furthermore, there are numerous studies that do not support engraftment by 23-fold. FISH revealed that engrafted male the plasticity of adult stem cells in different organs. Wagers et cells were localized to areas of bleomycin-induced injury and al. attempted to determine the fate of “prospectively isolated, exhibited an epithelium-like morphology, suggesting that stem long-term reconstituting HSCs” by using either a chimeric cells homed specifically to sites of injury. Furthermore, im- animal, produced via bone marrow ablation and reconstitution mediate administration of MSCs was able to protect the lung with GFP HSCs, or using a parabiotic model, joining the STEM CELLS FOR LUNG DISEASES 97R

Table 2. Characteristics of adult bone marrow-derived cells used in lung stem cell biology Cell type Phenotype Hematopoietic Stem Cells CD34ϩ/Ϫ, Stem cell antigen (SCA)-1ϩ, Thy1ϩ, c-kitϩ Lineage (lin)Ϫ—this is the absence of numerous markers for mature lymphocytes (CD45, CD3, CD4, CD8), myeloid (CD11b/Mac-1), and erythroid (TER-119) cells Mesenchymal Stem Cells no unique phenotype adherence to tissue culture plastic main isolation aid CD34ϩ linϪ MSCs (20) Plastic adherent MSCs (31) CD34ϩ linϪ MSCs (37) multi-potent adult (MAPC) (38) Side Population Cells (Hoechst dye efflux) (39) MSCs immunodepleted for CD34, CD45, and CD11b (40) MSCs immunodepleted for CD34, CD45, and CD11b (42) Total Bone Marrow, MSCs not isolated (46) Total Bone Marrow, and HSC side population cells (Hoechst dye efflux) (47)

circulation of a transgenic GFP mouse with the circulation of This result led to retrospectively analyze the lungs from mice a wild type mouse (45). While the BM ablated group showed receiving actin-eGFP bone marrow transplantation using de- robust reconstitution of circulating HSCs, and the parabiotic convolution microscopy. Deconvolution microscopy was able animals had robust hematopoietic chimerism, there was little to generate a three dimensional image of the lung allowing to evidence for stem cell engraftment and . determine that the co-expression of SP-C and eGFP was a Only one cell of 13.2 ϫ 106 cells examined within the brain, false positive: the pro-SP-C signal resided just outside the and seven cells of 4.7 ϫ 105 cells examined within the liver eGFPϩ cells, with a difference between the two signals less co-expressed the donor marker GFP as well as a tissue specific than 300 nm, and this phenomenon was not apparent until the marker (albumin for hepatocytes) in the BM ablation group three dimensional image was viewed from multiple angles (45). There was no evidence for engraftment and transdiffer- (46). entiation in the lung. Moreover, the parabiotic model had no Kotton et al. also used SP-C-eGFP mice, but investigated indication of HSC engraftment and transdifferentation in any both unfractionated BM as well as BM-derived SP cells (47). of the tissues analyzed (45). SP cells were purified from the femurs, tibias, and iliac crests In addition, two recent reports have challenged the engraft- and identified using a model of Hoechst dye efflux. These cells ment potential of adult-derived stem cells in the lung. These are enriched for HSC activity. Adult mice underwent two studies take advantage of transgenic mouse that express a myeloablation via either a single dose of 11 or 12 Gy radiation marker protein ubiquitously or under the control of the type II or two doses of 7 Gy (total of 14 Gy). The bone marrow was AEC specific promoter SP-C (46,47). Chang et al. trans- then reconstituted with either a whole bone marrow transplant planted bone marrow (2.0 ϫ 107 cells) from transgenic mice or transplantation of the highly enriched HSCs SP cells expressing the LacZ or eGFP gene ubiquitously (under the from SP-C-eGFP donor mice. Three months post-transplanta- control of the actin promoter), or under the control of the tion, with robust blood reconstitution, the mice were chal- human SP-C promoter into irradiated, neonatal mice. The lenged with a bleomycin injury (0.05 units) and engraftment lungs of recipients transplanted with bone marrow from trans- was assessed 1 mo post-transplant. Using three antibody genic mice that ubiquitously express eGFP or LacZ showed independent assays, fluorescent activated cell sorting cells whose morphology and location were compatible with (FACS), fluorescent microscopy, and real time PCR, the type II AEC (46). These green cells also co-localized with authors showed no evidence of donor cells becoming type II pro-SP-C. These initial results indicated that the eGFP BMSC AECs (47). were contributing to the alveolar epithelium. Flow cytometric In humans, BMSC repopulation also seems to contribute analysis of SP-C immunostained cells with eGFP indicated minimally to the type II AECs after cross-gender lung trans- that eGFP-fluorescent cells accounted for 50–55% of all lung plantation (49). Sequential immunohistochemistry and FISH cells, as well as 70% of type II AECs 2 wk after bone marrow performed on lung biopsy specimens from male recipients of transplantation (46). One month post-transplantation, eGFPϩ transplanted lungs from female donors showed Y-chromo- cells only comprised 2-25% of total lung cells with 0% to 8% some–containing type II AEC in 9 of 25 biopsy specimens of type II AECs being donor-derived. However, upon analysis from five of seven gender mismatched male lung transplant with cyto-centrifugation, the phenotype of the eGFPϩ SP-Cϩ recipients, that accounted for 0% to 0.553% of type II AEC cells displayed a small, round cell type with a scant cytoplasm (49). The number of type II AEC of male karyotype showed which is not congruent with the normal type II AEC pheno- a statistically significant relationship to the cumulative num- type of larger, cuboidal cells with an abundant cytoplasm (48). ber of episodes of acute cellular rejection. This study also Moreover, mice that received SP-C-eGFP bone marrow trans- suggests that BMSC contribute minimally to the type II AEC plant showed no eGFP expression, indicating that the trans- proliferation that is often present in these patients as a sequela planted cells are not adopting the type II AEC phenotype (46). to alveolar injury (49). 98R VAN HAAFTEN AND THE´ BAUD STEM CELL THERAPIES TO REPLACE DEFICIENT numbers of colony-forming units (CFUs) compared with con- GENES IN THE LUNG trol MSCs. ⌬F508 corrected MSCs showed improved chloride efflux in vitro. Furthermore, after co-culture with AECs, the The use of stem cells for CF is still at the conceptual stage, corrected ⌬F508 MSCs were able to secrete a significantly therefore there are currently very few reports on the use of larger amount of chloride when compared with uncorrected adult derived stem cells for CF. However, the idea that adult MSCs from ⌬F508 CF patients. Corrected ⌬F508 MSCs derived stem cells could be used as a vehicle for gene therapy responded to IBMX and forskolin (adenylate cyclase activa- is very appealing. Firstly, this method of administration would tors) to a larger degree than the uncorrected ⌬F508 MSCs give the physician a controlled method of administering the (52). While this in vitro proof of concept study is very corrected gene, circumventing some of the problems that have exciting, definitive answers cannot be concluded until this is been seen using viral mediated gene therapy (50). Secondly, a tested in vivo in an animal model of CF. stem cell could potentially provide a synergistic mechanism. There is one report to date attempting to use adult BMSC Not only would the stem cell deliver the corrected CFTR gene for CF in vivo. In this report, Loi et al. used both plastic to the lung of a CF patient, but these cells could also help adherent MSCs and total bone marrow harvested from trans- replenish destroyed tissue. genic mice over-expressing GFP (53). The authors subse- Grove et al. first showed the potential for a marrow derived quently ablated the bone marrow of CFTR knockout mice cell as a vehicle for gene delivery to the pulmonary epithelium using 800 rads total body irradiation and injected either the (51). MSCs were expanded in vitro after harvest from the bone MSCs (106 cells/mouse) or total bone marrow (20 ϫ 106 marrow by being grown in a defined basal bone marrow media cells/mouse) via the tail vein. They also used an intraperito- supplemented with human IL-6 (IL-6), murine IL-3, murine neal dose of naphthalene (275 mg/kg) either three days before stem cell factor, and polybrene. These cells were then trans- MSC administration or one month after bone marrow recon- duced with a retroviral vector (MND-eGFP-SN) that has been stitution with total bone marrow cells to induce lung injury optimized for long-term expression of eGFP in other cell lines. and stimulate migration of the transplanted bone marrow cells Following transduction, the eGFP marrow stem cells were to the lung. Stromal marrow cells were able to engraft into the then injected via the tail vein into mice that had received 600 alveolar epithelium and to confer CFTR mRNA to the knock- cGY of radiation for 2 consecutive days. This resulted in 1% out mice, but the engraftment occurred at such a low rate to 7% of AEC derived from donor eGFP stem cells (51). (0.025% chimeric airway epithelial cells, of which 0.01% More recently, the possibility of transforming in vitro expressed CFTR protein) that it is unlikely that this method MSCs from CF patients with a viral transcript to correct the would correct the trans-epithelial chloride deficit. Naphtha- defective CFTR gene was reported (52). MSCs were first lene-induced airway remodeling did not significantly increase obtained from healthy volunteers as well as CF volunteers the number of chimeric airway epithelial cells expressing with the ⌬F508 homozygous mutation. MSCs expressing CFTR. GFP, co-cultured with human airway epithelium, adopted a These two reports highlight the potential for BM-derived cuboidal or columnar shape similar to epithelial cells, as well stem cell based gene therapy in CF and the need for new as the expression of cytokeratin-18 (CK-18) a marker of strategies for enhancing airway epithelial engraftment to exert epithelial cells. When directly mixed with primary human a beneficial effect. AECs, control MSC expressed the tight junction protein oc- ϩ cludin. Moreover, FACS was used to isolate the GFP cells CONCLUSION from the co-culture system. These cells, when co-cultured with normal human AEC or with AEC from ⌬F508 homozy- The use of stem cells in the lung is at the crossroads of gous patients, expressed wild type CFTR, which were either understanding. Over the last decade, numerous studies have not detected in MSCs before or that were not co-cultured. demonstrated the ability of adult BM-derived cells to differ- These experiments strongly suggest that MSCs, when exposed entiate into non-hematopoietic lineages, including neural, to AECs, are able to adopt the phenotype of AECs and show skeletal muscle, hepatic, cardiac and vascular lineages and to the importance of the microenvironment in inducing tissue- repair these tissues. With regard to lung repair, the literature is specific transdifferentiation. While these results are very sparser. Initial promising reports showing adult BM-derived promising and lend credence to the ability of MSC transdif- cell homing, engraftment, and lung phenotype adoption are ferentiation into AEC, the true potential for MSC in CF are as now challenged. This highlights the need for more reliable vehicles for gene delivery. MSCs from 3 ⌬F508 homozygous techniques and assays to assess adult BM-derived cells lung patients were transduced with a viral vector for expression of engraftment and transdifferentiation. Other challenges before CFTR (vesicular stomatitis virus glycoprotein pseudotyped moving to the clinical application with this promising thera- Moloney murine virus-CFTR-neo). In this construct, peutic strategy include: improved characterization of putative the human CFTR gene was linked by internal ribosome entry BM-derived stem cells, determining mechanisms of homing, site to the neo gene to allow for selection of the corrected engraftment and transdifferentiation, as well as the alternate MSCs based on G418-resistance (a sulphate antibiotic) and mechanisms of action beyond pure tissue replacement, rigor- drug-resistant stem cell expansion. Insertion of the gene had ous assessment of therapeutic success combining lung histol- no effect on the multitpotency (based on adipogenic, chondro- ogy and function, and lastly and more importantly determining genic, and osteogenic cell differentiation), doubling times, and the long term safety of these MSCs. The potential for terato- STEM CELLS FOR LUNG DISEASES 99R mas and other cancers is too high a risk without certain 28. Kopen GC, Prockop DJ, Phinney DG 1999 Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into evidence that the exogenous administration of adult BM- neonatal mouse brains. Proc Natl Acad SciUSA96:10711–10716 derived stem cells is safe. 29. Kale S, Karihaloo A, Clark PR, Kashgarian M, Krause DS, Cantley LG 2003 Bone marrow stem cells contribute to repair of the ischemically injured renal tubule. J Clin Invest 112:42–49 30. Zhao YD, Courtman DW, Deng Y, Kugathasan L, Zhang Q, Stewart DJ 2005 Rescue REFERENCES of monocrotaline-induced pulmonary arterial hypertension using bone marrow- derived endothelial-like progenitor cells: efficacy of combined cell and eNOS gene 1. Abman SH 2001 Bronchopulmonary dysplasia: “a vascular hypothesis”. Am J therapy in established disease. Circ Res 96:442–450 Respir Crit Care Med 164:1755–1756 31. Kotton DN, Ma BY, Cardoso WV, Sanderson EA, Summer RS, Williams MC, Fine 2. Jobe AJ 1999 The new BPD: an arrest of lung development. Pediatr Res 46:641–643 A 2001 Bone marrow-derived cells as progenitors of lung alveolar epithelium. 3. Orenstein DM, Stern RC, Rosenstein BJ 2000 Cystic Fibrosis: Medical Care. Development 128:5181–5188 Lippincott Williams & Wilkins, Philadelphia, pp 1–21. 32. Jones AW, Reeve NL 1978 Ultrastructural study of bleomycin-induced pulmonary 4. Vankeerberghen A, Cuppens H, Cassiman JJ 2002 The cystic fibrosis transmem- changes in mice. J Pathol 124:227–233 brane conductance regulator: an intriguing protein with pleiotropic functions. J Cyst 33. Krane CM, Fortner CN, Hand AR, McGraw DW, Lorenz JN, Wert SE, Towne JE, Fibros 1:13–29 Paul RJ, Whitsett JA, Menon AG 2001 Aquaporin 5-deficient mouse lungs are 5. Evans MJ, Kaufman MH 1981 Establishment in culture of pluripotential cells from hyperresponsive to cholinergic stimulation. Proc Natl Acad SciUSA98:14114– mouse embryos. Nature 292:154–156 14119 6. McLaren A 2001 Ethical and social considerations of stem cell research. Nature 34. Rishi AK, Joyce-Brady M, Fisher J, Dobbs LG, Floros J, VanderSpek J, Brody JS, 414:129–131 Williams MC 1995 Cloning, characterization, and development expression of a rat 7. Hatch HM, Zheng D, Jorgensen ML, Petersen BE 2002 SDF-1alpha/CXCR4: a lung alveolar type I cell gene in embryonic endodermal and neural derivatives. Dev mechanism for hepatic oval cell activation and bone marrow stem cell recruitment to Biol 167:294–306 the injured liver of rats. Cloning Stem Cells 4:339–351 35. Williams MC 2003 Alveolar type I cells: molecular phenotype and development. 8. Michalopoulos GK, DeFrances MC 1997 Liver regeneration. Science 276:60–66 Annu Rev Physiol 65:669–695 9. Mason RJ, Williams MC, Moses HL, Mohla S, Berberich MA 1997 Stem cells in 36. Horowitz S, Watkins RH, Auten RL, Mercier CE, Cheng ER 1991 Differential lung development, disease, and therapy. Am J Respir Cell Mol Biol 16:355–363 accumulation of surfactant protein A, B, and C mRNAs in two epithelial cell types 10. Warburton D, Wuenschell C, Flores-Delgado G, Anderson K 1998 Commitment and of hyperoxic lung. Am J Respir Cell Mol Biol 5:511–515 differentiation of lung cell lineages. Biochem Cell Biol 76:971–995 37. Theise ND, Henegariu O, Grove J, Jagirdar J, Kao PN, Crawford JM, Badve S, 11. Pitt BR, Ortiz LA 2004 Stem cells in lung biology. Am J Physiol Lung Cell Mol Saxena R, Krause DS 2002 Radiation pneumonitis in mice: a severe injury model for Physiol 286:L621–L623 pneumocyte engraftment from bone marrow. Exp Hematol 30:1333–1338 12. Driscoll B, Buckley S, Bui KC, Anderson KD, Warburton D 2000 in 38. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez alveolar epithelial development and repair. Am J Physiol Lung Cell Mol Physiol XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low 279:L1191–L1198 WC, Largaespada DA, Verfaillie CM 2002 Pluripotency of mesenchymal stem cells 13. Reddy R, Buckley S, Doerken M, Barsky L, Weinberg K, Anderson KD, Warburton derived from adult marrow. Nature 418:41–49 D, Driscoll B 2004 Isolation of a putative progenitor subpopulation of alveolar 39. Abe S, Lauby G, Boyer C, Rennard SI, Sharp JG 2003 Transplanted BM and BM epithelial type 2 cells. Am J Physiol Lung Cell Mol Physiol 286:L658–L667 side population cells contribute progeny to the lung and liver in irradiated mice. 14. Wagers AJ, Weissman IL 2004 Plasticity of adult stem cells. Cell 116:639–648 Cytotherapy 5:523–533 15. Blau HM, Brazelton TR, Weimann JM 2001 The evolving concept of a stem cell: 40. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG entity or function? Cell 105:829–841 2003 Mesenchymal stem cell engraftment in lung is enhanced in response to 16. Friedenstein AJ, Gorskaja JF, Kulagina NN 1976 precursors in normal bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad SciUSA and irradiated mouse hematopoietic organs. Exp Hematol 4:267–274 100:8407–8411 17. Gronthos S, Zannettino AC, Hay SJ, Shi S, Graves SE, Kortesidis A, Simmons PJ 41. Vandervelde S, van Luyn MJ, Tio RA, Harmsen MC 2005 Signaling factors in stem 2003 Molecular and cellular characterisation of highly purified stromal stem cells cell-mediated repair of infarcted myocardium. J Mol Cell Cardiol 39:363–376 derived from human bone marrow. J Cell Sci 116:1827–1835 42. Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J, Brigham KL 2005 Bone 18. Jones EA, Kinsey SE, English A, Jones RA, Straszynski L, Meredith DM, Markham marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir AF, Jack A, Emery P, McGonagle D 2002 Isolation and characterization of bone Cell Mol Biol 33:145–152 marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 46:3349– 43. Wilson WL, Mullen M, Olley PM, Rabinovitch M 1985 Hyperoxia-induced pulmo- 3360 nary vascular and lung abnormalities in young rats and potential for recovery. Pediatr 19. Tropel P, Noel D, Platet N, Legrand P, Benabid AL, Berger F 2004 Isolation and Res 19:1059–1067 characterisation of mesenchymal stem cells from adult mouse bone marrow. Exp 44. Thebaud B, Ladha F, Michelakis ED, Sawicka M, Thurston G, Eaton F, Hashimoto Cell Res 295:395–406 K, Harry G, Haromy A, Korbutt G, Archer SL 2005 Vascular endothelial growth 20. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, Neutzel factor gene therapy increases survival, promotes lung angiogenesis, and prevents S, Sharkis SJ 2001 Multi-organ, multi-lineage engraftment by a single bone marrow- alveolar damage in hyperoxia-induced lung injury: evidence that angiogenesis derived stem Cell Cell 105:369–377 participates in alveolarization. Circulation 112:2477–2486 21. Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, Durand DJ, 45. Wagers AJ, Sherwood RI, Christensen JL, Weissman IL 2002 Little evidence for Fitton T, Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman developmental plasticity of adult hematopoietic stem cells. Science 297:2256–2259 AW, Hare JM 2005 Cardiac repair with intramyocardial injection of allogeneic 46. Chang JC, Summer R, Sun X, Fitzsimmons K, Fine A 2005 Evidence that bone mesenchymal stem cells after myocardial infarction. Proc Natl Acad SciUSA marrow cells do not contribute to the alveolar epithelium. Am J Respir Cell Mol Biol 102:11474–11479 33:335–342 22. Kajstura J, Rota M, Whang B, Cascapera S, Hosoda T, Bearzi C, Nurzynska D, 47. Kotton DN, Fabian AJ, Mulligan RC 2005 Failure of bone marrow to reconstitute Kasahara H, Zias E, Bonafe M, Nadal-Ginard B, Torella D, Nascimbene A, Quaini lung epithelium. Am J Respir Cell Mol Biol 33:328–334 F, Urbanek K, Leri A, Anversa P 2005 Bone marrow cells differentiate in cardiac cell 48. Shannon JM, Mason RJ, Jennings SD 1987 Functional differentiation of alveolar lineages after infarction independently of cell fusion. Circ Res 96:127–137 type II epithelial cells in vitro: effects of cell shape, cell-matrix interactions and 23. Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay cell-cell interactions. Biochim Biophys Acta 931:143–156 R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P 2001 Bone marrow cells 49. Zander DS, Baz MA, Cogle CR, Visner GA, Theise ND, Crawford JM 2005 Bone regenerate infarcted myocardium. Nature 410:701–705 marrow-derived stem-cell repopulation contributes minimally to the Type II pneu- 24. Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, mocyte pool in transplanted human lungs. Transplantation 80:206–212 Scadova H, Qin G, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW 2005 50. Davies JC, Alton EW 2005 Airway gene therapy. Adv Genet 54:291–314 Clonally expanded novel multipotent stem cells from human bone marrow regen- 51. Grove JE, Lutzko C, Priller J, Henegariu O, Theise ND, Kohn DB, Krause DS 2002 erate myocardium after myocardial infarction. J Clin Invest 115:326–338 Marrow-derived cells as vehicles for delivery of gene therapy to pulmonary epithe- 25. Jang YY, Collector MI, Baylin SB, Diehl AM, Sharkis SJ 2004 Hematopoietic stem lium. Am J Respir Cell Mol Biol 27:645–651 cells convert into liver cells within days without fusion. Nat Cell Biol 6:532–539 52. Wang G, Bunnell BA, Painter RG, Quiniones BC, Tom S, Lanson NA, Spees JL, 26. Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, Boggs Bertucci D, Peister A, Weiss DJ, Valentine VG, Prockop DJ, Kolls JK 2005 Adult SS, Greenberger JS, Goff JP 1999 Bone marrow as a potential source of hepatic oval stem cells from bone marrow stroma differentiate into airway epithelial cells: cells. Science 284:1168–1170 potential therapy for cystic fibrosis. Proc Natl Acad SciUSA102:186–191 27. Theise ND, Badve S, Saxena R, Henegariu O, Sell S, Crawford JM, Krause DS 2000 53. Loi R, Beckett T, Goncz KK, Suratt BT, Weiss DJ 2006 Limited Restoration of Derivation of hepatocytes from bone marrow cells in mice after radiation-induced cystic fibrosis lung epithelium in vivo with adult marrow derived cells. Am J Respir myeloablation. Hepatology 31:235–240 Crit Care Med 173:171–179